Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 55
Filtrar
1.
Mol Pharm ; 19(11): 3877-3883, 2022 11 07.
Artigo em Inglês | MEDLINE | ID: mdl-36018674

RESUMO

The innovation of cancer immunotherapy is improving the prognosis of colorectal cancer (CRC) in clinics. Nevertheless, due to tumor heterogeneity and complex underlying inhibitory mechanisms, the therapeutic response greatly varies among different patients. To optimize the clinical management of CRC patients, it is critical to develop novel approaches for response monitoring and prediction. In the current study, we developed a novel near-infrared fluorescence (NIRF) imaging probe (Cy5.5-ICOS mAb) targeting the inducible T-cell costimulatory receptor (ICOS or CD278) and assessed its capacity for the detection of ICOS+-activated T cells in vivo. ICOS expression was evaluated by flow cytometry and immunofluorescence staining in subcutaneous MC38 models treated with the stimulator of interferon genes (STING) agonist (STINGa). NIRF imaging study was performed 1 day after the last treatment, and tumor volume was monitored every other day with a caliper. A significantly higher optical signal could be detected at tumor regions in STINGa group, compared with that in the PBS group at all time points imaged, and this was in line with ex vivo imaging and immunofluorescence staining study. The data demonstrated that Cy5.5-ICOS mAb could detect ICOS+-activated T cells with high specificity, and ICOS NIRF imaging is a promising strategy for predicting and monitoring immune response in CRC.


Assuntos
Neoplasias Colorretais , Imunoterapia , Humanos , Carbocianinas , Imunoterapia/métodos , Diagnóstico por Imagem , Corantes Fluorescentes , Neoplasias Colorretais/diagnóstico por imagem , Neoplasias Colorretais/tratamento farmacológico
2.
Cancer Cell Int ; 19: 248, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31572065

RESUMO

BACKGROUND: Neuropilin-1 (NRP-1) is a non-tyrosine kinase receptor interacting with multiple signaling pathways that underpin the biological behavior and fate of cancer cells. However, in pancreatic cancer, the mechanisms underlying the function of NRP-1 in cell proliferation and metastasis and the involvement of regulatory upstream miRNAs remain unclear. METHODS: Potential miRNAs were mined by using multiple bioinformatics prediction tools and validated by luciferase assays. The expression of NRP-1 and miRNA-141 (miR-141) in pancreatic tissues and cells was examined by immunohistochemistry, immunoblotting and/or real-time RT-PCR. Stable transfected cells depleted of NRP-1 were generated, and regulatory effects of miR-141 were investigated by transfecting cells with miR-141 mimics and anti-miR-141. Assays of cell viability, proliferation, cell cycle distribution, transwell migration and cell scratch were employed. Xenograft tumor models were established to assess the effects of NRP-1 depletion on tumorigenesis and liver metastasis, and therapeutic effects of miR-141 on tumor growth. The role of miR-141/NRP-1 axis in regulating epithelial-mesenchymal transition (EMT) by co-interacting the TGF-ß pathway was examined. RESULTS: In this study, of 12 candidate miRNAs identified, miR-141 showed the strongest ability to regulate NRP-1. In pancreatic cancer tissues and cells, the expression level of NRP-1 was negatively correlated with that of miR-141. NRP-1 was highly expressed in pancreatic cancer tissues compared with normal pancreatic tissues, and its expression levels were positively correlated with tumor grade, lymph metastasis and AJCC staging. NRP-1 depletion inhibited cell proliferation by inducing cell cycle arrest at the G0/G1 phase through upregulating p27 and downregulating cyclin E and cyclin-dependent kinase 2, and reduced cell migration by inhibiting EMT through upregulating E-cadherin and downregulating Snail and N-cadherin. Through downregulating NRP-1, miR-141 mimics showed a similar effect as NRP-1 depletion on cell proliferation and migration. NRP-1 depletion suppressed tumor growth and liver metastasis and miR-141 mimics inhibited the growth of established tumors in mice. NRP-1 depletion and/or miR-141 mimics inhibited the activation of the TGF-ß pathway stimulated by TGF-ß ligand. CONCLUSIONS: The present results indicate that NRP-1 is negatively regulated by miR-141 and the miR-141/NRP-1 axis may serve as potentially valuable biomarkers and therapeutic targets for pancreatic cancer.

3.
Cell Physiol Biochem ; 47(3): 1025-1041, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29843141

RESUMO

BACKGROUND/AIMS: Recent evidence has shown that some long non-coding RNAs (lncRNAs) play important roles in various biological processes. However, the regulatory mechanism of lncRNA in gastric cancer (GC) remains unclear. METHODS: We reannotated the GC gene expression profile into a lncRNA-mRNA biphasic profile and integrated the microRNA target data to construct a global GC triple network. A further clustering and random walk with restart analyses was performed on the triple network from the level of topology analyses. Quantitative real-time PCR was used to determine expression of lncRNA RP11-363E7.4. Kaplan-Meier analyses was performed to evaluate the prognostic value of lncRNA RP11-363E7.4. RESULTS: We constructed a gastric cancer lncRNA-miRNA-mRNA network (GCLMN) including six lncRNAs, 332 mRNAs, and 3,707 edges. For the shared lncRNA RP11-363E7.4, the interacting gene and microRNA functional enrichment studies implied that lncRNA RP11-363E7.4 might function as a new regulator in GC. The expression of lncRNA RP11-363E7.4 was downregulated compared with that of paracarcinoma tissues in five GC samples. High expression of lncRNA RP11-363E7.4 was found to be correlated to better overall survival (OS) for GC patients. CONCLUSIONS: This study focused on GC lncRNA-miRNA-mRNA regulatory networks, and found that lncRNA RP11-363E7.4 was a new GC risk lncRNA, which might provide novel insight into a better understanding of the pathogenesis of GC.


Assuntos
Regulação para Baixo , Regulação Neoplásica da Expressão Gênica , MicroRNAs/biossíntese , RNA Longo não Codificante/biossíntese , RNA Mensageiro/biossíntese , RNA Neoplásico/biossíntese , Neoplasias Gástricas/metabolismo , Humanos , Neoplasias Gástricas/patologia
4.
Mol Carcinog ; 55(12): 2260-2267, 2016 12.
Artigo em Inglês | MEDLINE | ID: mdl-26800338

RESUMO

Colorectal cancer (CRC) is one of the leading causes of cancer death worldwide. MiR-382 has been found to have a decreased expression and the ability to suppress tumorigenesis in certain cancers. However, the role of miR-382 in CRC has not been sufficiently investigated. NR2F2 (also known as COUP-TFII), a member of the steroid/thyroid receptor superfamily, is often aberrantly activated in various tumors, but it is currently unclear whether NR2F2 may be a target of miR-382. In the present study, we investigated the role of miR-382 in CRC and identified the regulation of NR2F2 by miR-382. We observed that miR-382 was aberrantly downregulated in CRC. Transfection with miR-382 mimics impeded the growth, migration, and invasion of CRC cells. The direct binding of miR-382 to the 3' untranslated region (3' UTR) of NR2F2 was confirmed using a luciferase reporter gene assay. We showed that the relative expression levels of NR2F2 were significantly higher in CRC tissues compared with normal adjacent mucosa. A Kaplan-Meier analysis indicated that patients with high NR2F2 expression had a poor overall survival. Knockdown of NR2F2 inhibited CRC cell growth, migration, and invasion. Ectopic expression of NR2F2 mitigated miR-382 suppression of CRC cell proliferation, migration, and invasion. In conclusion, the present study describes a potential mechanism underlying a miR-382/NR2F2 link contributing to CRC development. Our results demonstrate that miR-382 represents a potential strategy against CRC. © 2016 Wiley Periodicals, Inc.


Assuntos
Fator II de Transcrição COUP/genética , Colo/patologia , Neoplasias Colorretais/genética , Regulação Neoplásica da Expressão Gênica , MicroRNAs/genética , Invasividade Neoplásica/genética , Reto/patologia , Linhagem Celular Tumoral , Movimento Celular , Proliferação de Células , Colo/metabolismo , Neoplasias Colorretais/patologia , Regulação para Baixo , Células HCT116 , Humanos , Invasividade Neoplásica/patologia , Reto/metabolismo
5.
IUBMB Life ; 68(3): 201-10, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-26825578

RESUMO

Colon cancer-associated transcript-1 (CCAT1) is a highly conserved long noncoding RNA that is deregulated in several cancers. However, its role in gastric carcinoma and its post-transcriptional regulation remain poorly understood. In this study, we provide the first evidence that CCAT1 regulates miR-490 in gastric cancer (GC) cells. Interestingly, miR-490 can also repress CCAT1 expression. CCAT1 expression was significantly upregulated, and miR-490 expression was downregulated in GC. The negative correlation between miR-490 and CCAT1 expression was observed in GC tissues. Importantly, CCAT1 contains a putative miR-490-binding site, and deletion of this binding site abolishes their miR-490 responsiveness. Post-transcriptional CCAT1 silencing by miR-490 significantly suppressed GC cell migration. Furthermore, miR-490 directly bound to the hnRNPA1 mRNA 3'-UTR to repress its translation. Inhibition of miR-490 rescued CCAT1 siRNA-mediated suppression of cell migration. hnRNPA1 expression was significantly upregulated in GC specimens, and there was a negative correlation between miR-490 and hnRNPA1 expression and also a positive correlation between hnRNAP1 expression level and CCAT1 level. Taken together, we show for the first time that the CCAT1/miR-490/hnRNPA1 axis promotes GC migration, and it may have a possible diagnostic and therapeutic potential in GC.


Assuntos
Movimento Celular , Ribonucleoproteínas Nucleares Heterogêneas Grupo A-B/genética , MicroRNAs/fisiologia , RNA Longo não Codificante/fisiologia , Neoplasias Gástricas/patologia , Sequência de Bases , Sítios de Ligação , Linhagem Celular Tumoral , Proliferação de Células , Feminino , Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Ribonucleoproteína Nuclear Heterogênea A1 , Ribonucleoproteínas Nucleares Heterogêneas Grupo A-B/metabolismo , Humanos , Masculino , Pessoa de Meia-Idade , Dados de Sequência Molecular , Interferência de RNA , Neoplasias Gástricas/metabolismo
6.
Tumour Biol ; 37(1): 641-51, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26242263

RESUMO

CCL18 is a member of CCL chemokines and is frequently overexpressed in cancer. Elevated CCL18 expression has been reported to be associated with poor prognosis of gastric cancer. However, the molecular mechanisms of CCL18 in gastric cancer cells remain elusive. In our study, we found that CCL18 was highly expressed in different gastric cancer cells. CCL18 stimulation dose-dependently enhanced the invasion and migration of MGC-803 cells. Knockdown of endogenous CCL18 inhibited the invasion and migration of MGC-803 cells, whereas overexpression of CCL18 promoted the invasion and migration of MKN28 cells. We further found that CCL18 increased the expressions of MMP-3 and Slug and decreased the expression of E-cadherin in MGC-803 cells. In addition, CCL18 time-dependently induced activation of ERK1/2, IκBα, and NF-κB. These effects of CCL18 were prevented by ERK1/2 selective inhibitor U0126 as well as NF-κB selective inhibitor BAY117082. Taken together, our findings establish a signaling role for CCL18 in gastric cancer cells and identify that the CCL18/ERK1/2/NF-κB signaling pathway is essential for tumor invasiveness in gastric cancer cells. Thus, our data may provide knowledge for using CCL18 as a novel target for effective diagnosis and treatment of gastric cancer.


Assuntos
Quimiocinas CC/metabolismo , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , NF-kappa B/metabolismo , Transdução de Sinais , Neoplasias Gástricas/metabolismo , Caderinas/genética , Caderinas/metabolismo , Linhagem Celular Tumoral , Movimento Celular , Quimiocinas CC/genética , Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Humanos , Metaloproteinase 3 da Matriz/genética , Metaloproteinase 3 da Matriz/metabolismo , Inibidores de Proteínas Quinases/farmacologia , Transdução de Sinais/efeitos dos fármacos , Fatores de Transcrição da Família Snail/genética , Fatores de Transcrição da Família Snail/metabolismo , Neoplasias Gástricas/genética , Neoplasias Gástricas/patologia
7.
Tumour Biol ; 37(1): 381-91, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26219898

RESUMO

Cabozantinib is a multi-targeted tyrosine kinase inhibitor targeting vascular endothelial growth factor (VEGF) receptor (VEGFR)-2, MET (c-Met, also called hepatocyte growth factor (HGF) receptor), and other receptor tyrosine kinases. Cabozantinib has recently been approved for treating advanced medullary thyroid carcinoma (MTC), but its long-term benefit remains uncertain and dose-dependent adverse events are very common. The present study has demonstrated that 2-methoxyestradiol (2ME2), an inhibitor of hypoxia-inducible factors (HIFs) and a promising anticancer agent under investigation in clinical trials, strengthens anticancer activities of cabozantinib against MTC cells in vitro and in vivo. The activated hypoxia-inducible pathways, which are mainly regulated by HIF-1, contribute to the resistance of hypoxic MTC cells to cabozantinib. Cabozantinib upregulated HIF-1α expression at translational levels and increased the expression of the downstream factors including VEGF, lactate dehydrogenase A (LDHA), HGF, and MET. 2ME2 corrected the activated pathways by cabozantinib through downregulating HIF-1α expression and inhibiting its nuclear translocation in hypoxic MTC cells. Administration of 2ME2 enhanced the efficacy of cabozantinib in suppressing the growth of MTC cell line xenografts and patient-derived xenografts established in mice. Given that 2ME2 targets insensitive hypoxic cancer cells to cabozantinib and can inhibit the activated pathways by cabozantinib, the present results warrant further investigation of 2ME2, particularly in combination with cabozantinib, for the treatment of MTC.


Assuntos
Anilidas/química , Carcinoma Neuroendócrino/metabolismo , Estradiol/análogos & derivados , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Piridinas/química , Neoplasias da Glândula Tireoide/metabolismo , 2-Metoxiestradiol , Transporte Ativo do Núcleo Celular , Adulto , Animais , Antineoplásicos/química , Apoptose , Carcinoma Neuroendócrino/tratamento farmacológico , Proliferação de Células , Sobrevivência Celular , Estradiol/química , Feminino , Humanos , Hipóxia , Isoenzimas/metabolismo , Antígeno Ki-67/metabolismo , L-Lactato Desidrogenase/metabolismo , Lactato Desidrogenase 5 , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Transplante de Neoplasias , Transdução de Sinais , Neoplasias da Glândula Tireoide/tratamento farmacológico , Ensaios Antitumorais Modelo de Xenoenxerto
8.
Hepatobiliary Pancreat Dis Int ; 15(2): 189-97, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-27020636

RESUMO

BACKGROUND: Most of the reports on the prognostic indicators of patients with pancreatic adenocarcinoma are from developed countries. The present study focused on the prognostic indicators of Chinese patients with pancreatic adenocarcinoma. METHODS: A total of 300 patients with pancreatic adenocarcinoma who had undergone curative resection were included. The resection and R0/R1 resection rates for adenocarcinomas from different parts of the pancreas were calculated and clinical characteristics were analyzed. RESULTS: In 3427 patients diagnosed with pancreatic adenocarcinomas, only 300 (8.8%) were eligible for radical resection. The total median survival of these patients was 19 months, and their 1-, 3-, and 5-year survival rates were 72.5%, 28.0% and 23.4%, respectively. The prognostic factors included socioeconomic status, smoking history, symptoms, high blood glucose, and various tumor characteristics, including perineural and vascular invasion, lymph node metastases, and CA19-9 levels before and after operation. Operation-associated prognostic indicators included operation time, blood loss and transfusions, pancreatic fistula, and complications. Independent predictors of mortality included poor socioeconomic status, smoking history, symptoms, CA19-9, perineural invasion and lymph node metastasis, grade of fistula and complications. Patient survival was not correlated with either resection margin or adjuvant chemotherapy in multivariate analysis. CONCLUSIONS: The survival rates of patients with curative resection for pancreatic adenocarcinoma in China are close to those in developed countries, but curative resection rate is far below. Socioeconomic status, symptoms, and CA19-9 are the three most prominent prognostic factors, which are helpful in patient selection and perioperative care.


Assuntos
Adenocarcinoma/cirurgia , Pancreatectomia , Neoplasias Pancreáticas/cirurgia , Adenocarcinoma/sangue , Adenocarcinoma/mortalidade , Adenocarcinoma/patologia , Antígeno CA-19-9/sangue , Distribuição de Qui-Quadrado , China , Feminino , Hospitais com Alto Volume de Atendimentos , Humanos , Estimativa de Kaplan-Meier , Masculino , Pessoa de Meia-Idade , Análise Multivariada , Pancreatectomia/efeitos adversos , Neoplasias Pancreáticas/sangue , Neoplasias Pancreáticas/mortalidade , Neoplasias Pancreáticas/patologia , Modelos de Riscos Proporcionais , Fatores de Risco , Fatores Socioeconômicos , Fatores de Tempo , Resultado do Tratamento
9.
J Vasc Res ; 51(4): 259-68, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25196016

RESUMO

OBJECTIVE: The present study aims to investigate the underlying mechanisms accounting for the activities of everolimus to inhibit the growth of vascular smooth muscle cells (VSMCs), which contributes to restenosis. METHODS: Primary VSMCs were cultured in media containing smooth muscle growth supplements and incubated with testing agents. Cell proliferation, cell cycle distribution, apoptosis and autophagy, and the key molecules involved, were examined. RESULTS: Everolimus inhibited the proliferation of VSMCs by inhibiting the activation of ribosomal protein S6 kinase and phosphorylation of eukaryotic translation initiation factor 4E-binding protein 1, and downregulating proliferating cellular nuclear antigen. Everolimus induced cell cycle arrest at the G1 phase by downregulating cyclin D1 and upregulating p27, and increased apoptosis by downregulating Bcl-2, upregulating Bad and activating capsase-3 and poly ADP ribose polymerase. Everolimus enhanced autophagy by increasing the conversion of microtubule-associated protein 1 light chain 3 (LC3)-I to LC3-II, and upregulating Beclin 1. Specific autophagy inhibitors, 3-methyladenine and bafilomycin A1, significantly attenuated the inhibition of cell proliferation, the increased apoptosis and the altered expression of the above key proteins induced by everolimus. CONCLUSIONS: Enhanced autophagy by everolimus contributes to its antirestenotic activity and its abilities to inhibit cell proliferation and to induce apoptosis of VSMCs.


Assuntos
Autofagia/efeitos dos fármacos , Músculo Liso Vascular/efeitos dos fármacos , Sirolimo/análogos & derivados , Resistência Vascular/efeitos dos fármacos , Animais , Aorta Torácica/citologia , Apoptose/efeitos dos fármacos , Apoptose/fisiologia , Autofagia/fisiologia , Proteínas de Transporte/metabolismo , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Ciclina D1/metabolismo , Inibidor de Quinase Dependente de Ciclina p27/metabolismo , Everolimo , Imunossupressores/farmacologia , Peptídeos e Proteínas de Sinalização Intracelular , Masculino , Músculo Liso Vascular/citologia , Músculo Liso Vascular/fisiologia , Fosfoproteínas/metabolismo , Antígeno Nuclear de Célula em Proliferação/metabolismo , RNA Interferente Pequeno/genética , Ratos Wistar , Proteínas Quinases S6 Ribossômicas/metabolismo , Sirolimo/farmacologia , Serina-Treonina Quinases TOR/antagonistas & inibidores , Resistência Vascular/fisiologia
10.
J Clin Gastroenterol ; 48(3): 261-3, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23842219

RESUMO

Solid evidence has revealed unsuspected functions of the spleen. Asplenic or hyposplenic patients bear an increased susceptibility to overwhelming postsplenectomy infection. We, for the first time, performed the split splenic transplantation to redress hemophilia A. We also resected the middle part of the spleen completely and anatomically for the first time. Our findings illustrate that the spleen can be divided into 2 functional parts, and may shed a new light on the understanding and practice in splenic surgery.


Assuntos
Hemangioma/cirurgia , Hemofilia A/cirurgia , Baço/irrigação sanguínea , Baço/transplante , Esplenectomia , Neoplasias Esplênicas/cirurgia , Adulto , Fator VIII/biossíntese , Feminino , Hemangioma/patologia , Hemofilia A/sangue , Hemofilia A/diagnóstico , Humanos , Imunossupressores/uso terapêutico , Doadores Vivos , Masculino , Baço/metabolismo , Neoplasias Esplênicas/patologia , Tomografia Computadorizada por Raios X , Resultado do Tratamento , Adulto Jovem
11.
Tumour Biol ; 34(1): 181-92, 2013 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-23229098

RESUMO

S-phase kinase-associated protein-2 (Skp2) is overexpressed in human cancers and associated with poor prognosis. Skp2 acts as an oncogenic protein by enhancing cancer cell growth and tumor metastasis. The present study has demonstrated that small hairpin RNA (shRNA)-mediated downregulation of Skp2 markedly inhibits the viability, proliferation, colony formation, migration, invasion, and apoptosis of human gastric cancer MGC803 cells, which express a high level of Skp2. In contrast, Skp2 shRNA had only a slight effect on the above properties of BGC823 cells, which express a low level of Skp2. In accord, knockdown of Skp2 suppressed the ability of MGC803 cells to form tumors and to metastasize to the lungs of mice, and the growth of established tumors, by inhibiting cell proliferation and enhancing cell apoptosis. In contrast, overexpression of Skp2 promoted tumorigenesis of BGC823 cells in mice. Skp2 depletion induced cell cycle arrest in the G(1)/S phase by upregulating p27, p21, and p57 and downregulating cyclin E and cyclin-dependent kinase 2. Skp2 depletion also increased caspase-3 activity, impeded the ability of cells to form filopoidia and locomote, upregulated RECK (reversion-inducing cysteine-rich protein with kazal motifs), and downregulated matrix metalloproteinase (MMP)-2 and MMP-9 activity and expression. The results suggest that downregulating Skp2 warrants investigation as a promising strategy to treat gastric cancers that express high levels of Skp2.


Assuntos
Proteínas Quinases Associadas a Fase S/metabolismo , Neoplasias Gástricas/metabolismo , Neoplasias Gástricas/patologia , Animais , Apoptose , Caspase 3/biossíntese , Caspase 3/metabolismo , Pontos de Checagem do Ciclo Celular , Linhagem Celular Tumoral , Movimento Celular , Proliferação de Células , Ciclina E/biossíntese , Quinase 2 Dependente de Ciclina/biossíntese , Inibidor de Quinase Dependente de Ciclina p21/biossíntese , Inibidor de Quinase Dependente de Ciclina p27/biossíntese , Inibidor de Quinase Dependente de Ciclina p57/biossíntese , Regulação para Baixo , Proteínas Ligadas por GPI/biossíntese , Humanos , Masculino , Metaloproteinase 2 da Matriz/biossíntese , Metaloproteinase 9 da Matriz/biossíntese , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Invasividade Neoplásica , Metástase Neoplásica , Interferência de RNA , RNA Interferente Pequeno , Proteínas Quinases Associadas a Fase S/genética , Neoplasias Gástricas/genética , Regulação para Cima
12.
Cancer Sci ; 103(3): 528-34, 2012 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-22145922

RESUMO

The hypoxic microenvironment inside solid tumors, including hepatocellular carcinoma (HCC), is a major cause of tumor resistance to chemotherapy. The recently identified hypoxia-inducible factor (HIF)-2 executes the hypoxia response. Its expression feature and transcriptional targets indicate a possible dominance of HIF-2 in regulating genes in HCC. The aim of the present study was to determine whether transfection of siRNA targeting HIF-2α could enhance the efficacy of doxorubicin, the most commonly used drug in the treatment of HCC. Transfection of HIF-2 siRNA into human HCC cells downregulated the expression of HIF-2α, vascular endothelial growth factor (VEGF), transforming growth factor (TGF)-α, and cyclin D1, but had little effect on the expression of HIF-1α, fms-related tyrosine kinase-1 (Flt-1), the glucose transporter (GLUT)-1, and lactate dehydrogenase A (LDHA). Doxorubicin itself only downregulated VEGF expression. Furthermore, HIF-2 siRNA inhibited proliferation, induced cell cycle arrest at the G(0)/G(1) phase, and acted synergistically with doxorubicin to inhibit the growth of human HCC cells in vitro. Transfection of HIF-2 siRNA also downregulated tumoral expression of HIF-2α, VEGF, TGF-α, and cyclin D1 in vivo, and acted synergistically with doxorubicin to suppress the growth of HepG2 tumors established in immunodeficient mice by inhibiting cell proliferation, tumor angiogenesis and microvessel perfusion. The results of the present study suggest that targeting HIF-2α with siRNA warrants investigation as a potential strategy to enhance the efficacy of doxorubicin in the treatment of HCC.


Assuntos
Antineoplásicos/uso terapêutico , Fatores de Transcrição Hélice-Alça-Hélice Básicos/antagonistas & inibidores , Carcinoma Hepatocelular/terapia , Doxorrubicina/uso terapêutico , Terapia Genética/métodos , Neoplasias Hepáticas/terapia , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Western Blotting , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/metabolismo , Linhagem Celular Tumoral , Regulação para Baixo , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Imuno-Histoquímica , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/metabolismo , Masculino , Camundongos , Camundongos Nus , RNA Interferente Pequeno/uso terapêutico , Reação em Cadeia da Polimerase em Tempo Real , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transfecção
13.
Invest New Drugs ; 30(2): 508-17, 2012 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-21080209

RESUMO

Liver metastasis is the major obstacle for prolonging the survival of colon cancer patients. Low-molecular-weight heparin (LMWH), a common drug for venous thromboembolism, has displayed beneficial effects in improving the survival of cancer patients, though the mechanism remains unclear. This study aimed to investigate the effects of LMWH on hepatic metastasis of colon cancer and its underlying molecular mechanism by targeting the interaction of the chemokine receptor CXCR4 and its ligand CXCL12 (formerly known as stromal cell-derived factor 1α, SDF-1α), as the CXCR4-CXCL12 axis has been shown to regulate the interaction of cancer cells and stroma. Experimental results revealed that LMWH (Enoxaparin, 3500-5500 Da) inhibited the CXCL12-stimulated proliferation, adhesion and colony formation of human colon cancer HCT-116 cells that highly expressed CXCR4. Interestingly, LMWH or an anti-CXCR4 blocking antibody diminished the migrating and invading abilities of HCT116 cells stimulated by the recombinant CXCL12 protein or liver homogenates which contained endogenous CXCL12 protein. Although LMWH did not significantly inhibit the growth of subcutaneous colon tumors, it significantly suppressed the formation of hepatic metastasis established by intrasplenic injection of colon cancer cells in nude Balb/c mice and also downregulated the expression of CXCL12 in hepatic sinusoidal endothelial cells. The results suggest that LMWH inhibits the formation of hepatic metastasis of colon cancer by disrupting the interaction of CXCR4 and CXCL12, supporting that perioperative administration of LMWH may help to prevent the seeding and subsequent growth of hepatic metastases of colon cancer cells.


Assuntos
Antineoplásicos/farmacologia , Quimiocina CXCL12/metabolismo , Neoplasias Colorretais/tratamento farmacológico , Células Endoteliais/efeitos dos fármacos , Enoxaparina/farmacologia , Neoplasias Hepáticas/irrigação sanguínea , Neoplasias Hepáticas/prevenção & controle , Receptores CXCR4/metabolismo , Animais , Adesão Celular/efeitos dos fármacos , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Neoplasias Colorretais/imunologia , Neoplasias Colorretais/patologia , Relação Dose-Resposta a Droga , Células Endoteliais/imunologia , Células Endoteliais/patologia , Células HCT116 , Células HT29 , Humanos , Neoplasias Hepáticas/imunologia , Neoplasias Hepáticas/secundário , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Invasividade Neoplásica , Fatores de Tempo , Ensaios Antitumorais Modelo de Xenoenxerto
14.
J Surg Res ; 166(2): e187-94, 2011 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-21227452

RESUMO

BACKGROUND: Heme oxygenase (HO)-1 protects transplanted organs from ischemia reperfusion injury and immune rejection. This study sought to investigate whether persistent overexpression of HO-1 in donor livers could improve the survival by expanding T regulatory cells in a rat model of orthotopic liver transplantation. METHODS: Livers of Dark Agouti rats were intraportally perfused with an AAV expression vector encoding rat HO-1 (AAV-HO-1), and then transplanted into Lewis rats. The survival, HO-1 activity, Banff rejection activity index, serum levels of IL-2 and TNF-α, infiltration of CD4(+), CD8(+), and T(reg) (CD4(+)CD25(+)Foxp3(+)) cells into donor livers, and expression of Foxp3, TGF-ß, and IL-10 were examined. A mixed lymphocyte reaction (MLR) was performed. RESULTS: Intraportal delivery of AAV-HO-1 resulted in persistent expression of HO-1 and increased activity of HO-1 in transplanted livers, leading to prolonged survival of recipients. Overexpression of HO-1 reduced the Banff rejection activity index, and production of IL-2 and TNF-α, inhibited infiltration of CD4(+) and CD8(+) cells, and increased infiltration of T(reg) cells, into donor livers. The spleens of recipients expressed higher levels of Foxp3, TGF-ß, and IL-10 than those of control rats, and the transplanted livers expressed higher levels of Foxp3 and TGF-ß. Splenocytes from the tolerant recipients had higher percentages of T(reg) cells, and responded poorly to the allogeneic donor splenocytes. CONCLUSIONS: Persistent expression of HO-1 in donor livers by intraportal delivery of AAV-HO-1 improves the survival by expanding T(reg) cells. HO-1-based therapies, as described herein, promise new strategies to prevent the rejection of liver transplants.


Assuntos
Terapia Genética/métodos , Sobrevivência de Enxerto/imunologia , Heme Oxigenase (Desciclizante)/genética , Transplante de Fígado , Traumatismo por Reperfusão/terapia , Linfócitos T Reguladores/citologia , Adenoviridae/genética , Animais , Linfócitos T CD8-Positivos/citologia , Linfócitos T CD8-Positivos/imunologia , Fatores de Transcrição Forkhead/genética , Rejeição de Enxerto/imunologia , Rejeição de Enxerto/prevenção & controle , Rejeição de Enxerto/terapia , Heme Oxigenase (Desciclizante)/imunologia , Interleucina-10/genética , Fígado/enzimologia , Fígado/imunologia , Fígado/cirurgia , Masculino , Ratos , Ratos Endogâmicos Lew , Ratos Wistar , Traumatismo por Reperfusão/imunologia , Traumatismo por Reperfusão/prevenção & controle , Baço/fisiologia , Linfócitos T Reguladores/imunologia , Fator de Crescimento Transformador beta/genética , Condicionamento Pré-Transplante , Transplante Homólogo
15.
Clin Dev Immunol ; 2011: 695834, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-22013483

RESUMO

B7-H4 is one of the most recently identified members of B7 superfamily of costimulatory molecules serving as an inhibitory modulator of T-cell response. B7-H4 is broadly expressed in human peripheral tissues and inducibly expressed in immune cells. The expression of B7-H4 has been observed in various types of human cancer tissues, and its soluble form has been detected in blood samples from cancer patients. However, its precise physiological role is still elusive, as its receptor has not been identified and the expression levels are not consistent. This paper summarizes the pertinent data on the inhibitory role of B7-H4 in antitumor immunity and its association with cancer progression and survival in human patients. The paper also discusses the clinical significance of investigating B7-H4 as potential markers for cancer diagnosis and prognosis, and as therapeutic targets.


Assuntos
Biomarcadores Tumorais/metabolismo , Terapia de Imunossupressão , Neoplasias/diagnóstico , Neoplasias/imunologia , Inibidor 1 da Ativação de Células T com Domínio V-Set/metabolismo , Antígenos de Neoplasias/imunologia , Transformação Celular Neoplásica , Progressão da Doença , Regulação Neoplásica da Expressão Gênica/imunologia , Humanos , Imunidade , Linfócitos do Interstício Tumoral/imunologia , Neoplasias/genética , Neoplasias/mortalidade , Prognóstico , Análise de Sobrevida , Evasão Tumoral , Microambiente Tumoral , Inibidor 1 da Ativação de Células T com Domínio V-Set/genética , Inibidor 1 da Ativação de Células T com Domínio V-Set/imunologia
16.
Surg Today ; 41(8): 1075-84, 2011 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-21773896

RESUMO

PURPOSE: Hepatic ischemia/reperfusion (HIR) injury is an unavoidable consequence of major liver surgery, during which endotoxemia often takes place. This study aimed to investigate whether matrine has a protective effect against HIR-induced liver injury aggravated by endotoxin. METHODS: Wistar rats were subjected to 30 min of HIR followed by lipopolysaccharide (LPS) (0.5 mg/kg) administration. At the indicated time points, six rats from each group (24 rats) were randomly euthanized to collect blood samples and livers. RESULTS: Preadministration of matrine protected livers from injury induced by HIR+LPS as the histological score, myeloperoxidase activity and malondialdehyde contents, expression of macrophage-inflammatory protein-2, DNA-binding activity of nuclear factor κB, and serum levels of alanine aminotransferase, aspartate aminotransferase, lactate dehydrogenase, alkaline phosphatase, tumor necrosis factor-α, soluble intercellular adhesion molecule-1, and nitric oxide were significantly reduced, and serum levels of interleukin-6 were further increased. HIR+LPS markedly induced cell apoptosis and necrosis, and upregulated the expression of cleaved caspase-3, Fas, and FasL. Matrine significantly reduced cell necrosis, but had a nonsignificant inhibitory effect on cell apoptosis and expression of cleaved caspase-3 and FasL. CONCLUSIONS: Matrine attenuates endotoxin-induced acute liver injury after HIR mainly by its anti-inflammatory and antioxidative activities, and has little inhibitory effect on cell apoptosis.


Assuntos
Alcaloides/uso terapêutico , Endotoxemia/prevenção & controle , Lipopolissacarídeos , Hepatopatias/prevenção & controle , Quinolizinas/uso terapêutico , Traumatismo por Reperfusão/prevenção & controle , Animais , Modelos Animais de Doenças , Endotoxemia/etiologia , Endotoxemia/patologia , Hepatopatias/etiologia , Hepatopatias/patologia , Masculino , Ratos , Ratos Wistar , Traumatismo por Reperfusão/etiologia , Traumatismo por Reperfusão/patologia , Matrinas
17.
Front Cell Dev Biol ; 9: 789587, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34901030

RESUMO

Transcriptomes and DNA methylation of colon cancer at the single-cell level are used to identify marker genes and improve diagnoses and therapies. Seven colon cancer subtypes are recognized based on the single-cell RNA sequence, and the differentially expressed genes regulated by dysregulated methylation are identified as marker genes for different types of colon cancer. Compared with normal colon cells, marker genes of different types show very obvious specificity, especially upregulated genes in tumors. Functional enrichment analysis for marker genes indicates a possible relation between colon cancer and nervous system disease, moreover, the weak immune system is verified in colon cancer. The heightened expression of markers and the reduction of methylation in colon cancer promote tumor development in an extensive mechanism so that there is no biological process that can be enriched in different types.

18.
Amino Acids ; 38(1): 237-45, 2010 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-19263195

RESUMO

Hepatic ischemia reperfusion (HIR) not only results in liver injury, but also leads to endotoxemia, which aggravates HIR-induced liver injury and dysfunction, or even causes liver failure. Taurine has been shown to protect organs from ischemia reperfusion or endotoxin by its anti-oxidant and anti-inflammatory activities. The aim of this study was to investigate whether taurine could attenuate endotoxin-induced acute liver injury after HIR. Wistar rats subjected to 30 min of hepatic ischemia followed by reperfusion and lipopolysaccharide (LPS) (0.5 mg/kg) administration, exhibited liver dysfunction (elevated serum levels of ALT, AST and LDH) and hepatic histopathological alteration. The serum levels of TNF-alpha and production of myeloperoxidase (MPO) and malondialdehyde (MDA) in liver tissues and apoptosis of hepatocytes were also increased after the combination of HIR and LPS. However, pre-administration of taurine protected livers from injury induced by the combination of HIR + LPS as the histological score, apoptotic index, MPO activity and production of MDA in liver tissues, and serum levels of AST, ALT, LDH and TNF-alpha, were significantly reduced. The expression of caspase-3, Fas and Fas ligand was upregulated in homogenates of livers from rats subjected to HIR and LPS, and this elevated expression could be inhibited by taurine. In summary, the results further emphasize the potential utilization of taurine in protecting livers against endotoxin-induced injury especially after HIR, by its anti-inflammatory, anti-oxidative and anti-apoptotic activities.


Assuntos
Endotoxinas/toxicidade , Fígado/efeitos dos fármacos , Fígado/lesões , Substâncias Protetoras/administração & dosagem , Traumatismo por Reperfusão/tratamento farmacológico , Taurina/administração & dosagem , Animais , Caspase 3/metabolismo , Injeções Intravenosas , L-Lactato Desidrogenase/metabolismo , Fígado/irrigação sanguínea , Fígado/enzimologia , Masculino , Malondialdeído/metabolismo , Peroxidase/metabolismo , Ratos , Traumatismo por Reperfusão/metabolismo
19.
Cancer Sci ; 100(11): 2226-33, 2009 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-19709125

RESUMO

Hepatocellular carcinoma (HCC) is one of the most common cancer-related causes of death, and conventional treatments offer unsatisfactory response. We have previously reported that kallistatin gene therapy suppressed the growth of HCC tumors by its anti-angiogenic activity, and meloxicam, a selective COX-2 inhibitor, inhibited proliferation and induced apoptosis of human HCC cells in vitro. The aim of this study was to determine whether combining kallistatin gene therapy and meloxicam could offer a better therapeutic effect to combat HCC in mice. A kallistatin expression plasmid was constructed and its expression was detected after intratumoral gene transfer. Both kallistatin gene therapy and meloxicam suppressed the growth of subcutaneous human HepG2 tumors established in BALB/c nude mice, and the combinational therapy showed a stronger effect in suppressing tumor growth, tumor angiogenesis and cell proliferation, and increasing cell apoptosis, than the respective monotherapies. Gene transfer of kallistatin inhibited tumor angiogenesis, and slightly inhibited cell proliferation and increased cell apoptosis in situ, but had no effect on expression of vascular endothelial growth factor, basic fibroblast growth factor, proliferating cell nuclear antigen, Bcl-2, Bax, or activation of caspase-3. Meloxicam therapy inhibited cell proliferation, induced cell apoptosis, reduced expression of proliferating cell nuclear antigen, increased activation of caspase-3, and upregulated Bax. Meloxicam also slightly inhibited tumor angiogenesis with no effect on the expression of vascular endothelial growth factor or basic fibroblast growth factor. Combining two novel anticancer agents, kallistatin targeting tumoral vascularization and meloxicam targeting cell proliferation and apoptosis, warrants investigation as a therapeutic strategy to combat HCC.


Assuntos
Inibidores de Ciclo-Oxigenase 2/uso terapêutico , Terapia Genética , Neoplasias Hepáticas Experimentais/terapia , Serpinas/genética , Tiazinas/uso terapêutico , Tiazóis/uso terapêutico , Animais , Apoptose/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Terapia Combinada , Neoplasias Hepáticas Experimentais/irrigação sanguínea , Neoplasias Hepáticas Experimentais/patologia , Masculino , Meloxicam , Camundongos , Camundongos Endogâmicos BALB C , Fator A de Crescimento do Endotélio Vascular/antagonistas & inibidores
20.
J Biomed Sci ; 16: 33, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-19292890

RESUMO

Developmentally regulated endothelial cell locus-1 (Del1) is an embryonic angiogenic factor expressed in early embryonic endothelial cells, but recently has been found to be expressed in some forms of cancers including colon and breast cancers, and melanoma, and human cancer cell lines. Overexpression of Del1 accelerates tumor growth by enhancing vascular formation, implying Del1 may be a potential target for anti-angiogenic cancer therapy. The study aims to investigate whether downregulation of Del1 could inhibit the growth of tumors established in nude Balb/c mice by subcutaneous implantation of human LS-174T colon cancer cells. The shRNA expression vectors targeting human Del1, and vascular endothelial growth factor (VEGF) were constructed. Gene transfection of Del1-shRNA downregulated expression of Del1 in LS-174T cells in vivo and in vitro, but did not alter the proliferative or survival properties of cells in vitro. Gene transfection of VEGF-shRNA downregulated expression of both VEGF and Del1 in LS-174T cells in vivo and in vitro. Both Del1-shRNA and VEGF-shRNA gene therapies exhibited anti-tumor activities and they also showed a synergistic effect in suppressing growth of colon tumors by anti-angiogenesis and anti-proliferation. Although further investigation to clarify the mechanisms explaining the role of Del1 in tumor growth, and the interaction between VEGF and Del1, is required, the results indicate that downregulation of Del1 presents a potent therapeutic strategy to combat colon cancer.


Assuntos
Proteínas de Transporte/biossíntese , Neoplasias do Colo/metabolismo , Regulação para Baixo , Regulação Neoplásica da Expressão Gênica , Proteínas de Neoplasias/biossíntese , Animais , Proteínas de Ligação ao Cálcio , Proteínas de Transporte/genética , Moléculas de Adesão Celular , Linhagem Celular Tumoral , Neoplasias do Colo/genética , Neoplasias do Colo/patologia , Neoplasias do Colo/terapia , Terapia Genética , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Proteínas de Neoplasias/genética , Neovascularização Patológica/genética , Neovascularização Patológica/metabolismo , Neovascularização Patológica/patologia , Neovascularização Patológica/terapia , Transfecção , Fator A de Crescimento do Endotélio Vascular/biossíntese , Fator A de Crescimento do Endotélio Vascular/genética
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa